Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 756
Filtrer
1.
Int J Mol Sci ; 25(13)2024 Jul 07.
Article de Anglais | MEDLINE | ID: mdl-39000557

RÉSUMÉ

The effects of intestinal microflora on extraintestinal immune response by intestinal cytokines and metabolites have been documented, but whether intestinal microbes stimulate serum antibody generation is unknown. Here, serum antibodies against 69 outer membrane proteins of Escherichia coli, a dominant bacterium in the human intestine, are detected in 141 healthy individuals of varying ages. Antibodies against E. coli outer membrane proteins are determined in all serum samples tested, and frequencies of antibodies to five outer membrane proteins (OmpA, OmpX, TsX, HlpA, and FepA) are close to 100%. Serum antibodies against E. coli outer membrane proteins are further validated by Western blot and bacterial pull-down. Moreover, the present study shows that OstA, HlpA, Tsx, NlpB, OmpC, YfcU, and OmpA provide specific immune protection against pathogenic E. coli, while HlpA and OmpA also exhibit cross-protection against Staphylococcus aureus infection. These finding indicate that intestinal E. coli activate extraintestinal antibody responses and provide anti-infective immunity.


Sujet(s)
Anticorps antibactériens , Protéines de la membrane externe bactérienne , Escherichia coli , Humains , Escherichia coli/immunologie , Anticorps antibactériens/immunologie , Anticorps antibactériens/sang , Protéines de la membrane externe bactérienne/immunologie , Adulte , Femelle , Staphylococcus aureus/immunologie , Mâle , Production d'anticorps/immunologie , Adulte d'âge moyen , Protéines Escherichia coli/immunologie , Jeune adulte , Sujet âgé , Infections à staphylocoques/immunologie , Infections à staphylocoques/microbiologie , Adolescent , Infections à Escherichia coli/immunologie , Infections à Escherichia coli/microbiologie
2.
PeerJ ; 12: e17336, 2024.
Article de Anglais | MEDLINE | ID: mdl-38784397

RÉSUMÉ

Background: Urinary tract infections (UTIs) are very common worldwide. According to their symptomatology, these infections are classified as pyelonephritis, cystitis, or asymptomatic bacteriuria (AB). Approximately 75-95% of UTIs are caused by uropathogenic Escherichia coli (UPEC), which is an extraintestinal bacterium that possesses virulence factors for bacterial adherence and invasion in the urinary tract. In addition, UPEC possesses type 6 secretion systems (T6SS) as virulence mechanisms that can participate in bacterial competition and in bacterial pathogenicity. UPEC UMN026 carries three genes, namely, ECUMN_0231, ECUMN_0232, and ECUMN_0233, which encode three uncharacterized proteins related to the T6SS that are conserved in strains from phylogroups B2 and D and have been proposed as biomarkers of UTIs. Aim: To analyze the frequency of the ECUMN_0231, ECUMN_0232, ECUMN_0233, and vgrG genes in UTI isolates, as well as their expression in Luria Bertani (LB) medium and urine; to determine whether these genes are related to UTI symptoms or bacterial competence and to identify functional domains on the putative proteins. Methods: The frequency of the ECUMN and vgrG genes in 99 clinical isolates from UPEC was determined by endpoint PCR. The relationship between gene presence and UTI symptomatology was determined using the chi2 test, with p < 0.05 considered to indicate statistical significance. The expression of the three ECUMN genes and vgrG was analyzed by RT-PCR. The antibacterial activity of strain UMN026 was determined by bacterial competence assays. The identification of functional domains and the docking were performed using bioinformatic tools. Results: The ECUMN genes are conserved in 33.3% of clinical isolates from patients with symptomatic and asymptomatic UTIs and have no relationship with UTI symptomatology. Of the ECUMN+ isolates, only five (15.15%, 5/33) had the three ECUMN and vgrG genes. These genes were expressed in LB broth and urine in UPEC UMN026 but not in all the clinical isolates. Strain UMN026 had antibacterial activity against UPEC clinical isolate 4014 (ECUMN-) and E. faecalis but not against isolate 4012 (ECUMN+). Bioinformatics analysis suggested that the ECUMN genes encode a chaperone/effector/immunity system. Conclusions: The ECUMN genes are conserved in clinical isolates from symptomatic and asymptomatic patients and are not related to UTI symptoms. However, these genes encode a putative chaperone/effector/immunity system that seems to be involved in the antibacterial activity of strain UMN026.


Sujet(s)
Infections à Escherichia coli , Protéines Escherichia coli , Chaperons moléculaires , Infections urinaires , Escherichia coli uropathogène , Escherichia coli uropathogène/immunologie , Escherichia coli uropathogène/génétique , Escherichia coli uropathogène/pathogénicité , Humains , Infections urinaires/microbiologie , Infections urinaires/immunologie , Chaperons moléculaires/génétique , Chaperons moléculaires/métabolisme , Infections à Escherichia coli/immunologie , Infections à Escherichia coli/microbiologie , Protéines Escherichia coli/génétique , Protéines Escherichia coli/immunologie , Protéines Escherichia coli/métabolisme , Femelle , Facteurs de virulence/génétique , Facteurs de virulence/immunologie , Mâle , Adulte d'âge moyen , Adulte
3.
An Acad Bras Cienc ; 96(2): e20231208, 2024.
Article de Anglais | MEDLINE | ID: mdl-38747841

RÉSUMÉ

The enterotoxigenic Escherichia coli (ETEC) strain is one of the most frequent causative agents of childhood diarrhea and travelers' diarrhea in low-and middle-income countries. Among the virulence factors secreted by ETEC, the exoprotein EtpA has been described as an important. In the present study, a new detection tool for enterotoxigenic E. coli bacteria using the EtpA protein was developed. Initially, antigenic sequences of the EtpA protein were selected via in silico prediction. A chimeric recombinant protein, corresponding to the selected regions, was expressed in an E. coli host, purified and used for the immunization of mice. The specific recognition of anti-EtpA IgG antibodies generated was evaluated using flow cytometry. The tests demonstrated that the antibodiesdeveloped were able to recognize the native EtpA protein. By coupling these antibodies to magnetic beads for the capture and detection of ETEC isolates, cytometric analyses showed an increase in sensitivity, specificity and the effectiveness of the method of separation and detection of these pathogens. This is the first report of the use of this methodology for ETEC separation. Future trials may indicate their potential use for isolating these and other pathogens in clinical samples, thus accelerating the diagnosis and treatment of diseases.


Sujet(s)
Anticorps antibactériens , Escherichia coli entérotoxigène , Protéines Escherichia coli , Cytométrie en flux , Animaux , Femelle , Souris , Anticorps antibactériens/immunologie , Escherichia coli entérotoxigène/immunologie , Protéines Escherichia coli/immunologie , Cytométrie en flux/méthodes , Immunoglobuline G/immunologie , Souris de lignée BALB C , Sensibilité et spécificité , Adhésines bactériennes/immunologie
4.
Infect Immun ; 92(5): e0044023, 2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38591882

RÉSUMÉ

Extraintestinal pathogenic Escherichia coli (ExPEC) is a leading cause of worldwide morbidity and mortality, the top cause of antimicrobial-resistant (AMR) infections, and the most frequent cause of life-threatening sepsis and urinary tract infections (UTI) in adults. The development of an effective and universal vaccine is complicated by this pathogen's pan-genome, its ability to mix and match virulence factors and AMR genes via horizontal gene transfer, an inability to decipher commensal from pathogens, and its intimate association and co-evolution with mammals. Using a pan virulome analysis of >20,000 sequenced E. coli strains, we identified the secreted cytolysin α-hemolysin (HlyA) as a high priority target for vaccine exploration studies. We demonstrate that a catalytically inactive pure form of HlyA, expressed in an autologous host using its own secretion system, is highly immunogenic in a murine host, protects against several forms of ExPEC infection (including lethal bacteremia), and significantly lowers bacterial burdens in multiple organ systems. Interestingly, the combination of a previously reported autotransporter (SinH) with HlyA was notably effective, inducing near complete protection against lethal challenge, including commonly used infection strains ST73 (CFT073) and ST95 (UTI89), as well as a mixture of 10 of the most highly virulent sequence types and strains from our clinical collection. Both HlyA and HlyA-SinH combinations also afforded some protection against UTI89 colonization in a murine UTI model. These findings suggest recombinant, inactive hemolysin and/or its combination with SinH warrant investigation in the development of an E. coli vaccine against invasive disease.


Sujet(s)
Infections à Escherichia coli , Protéines Escherichia coli , Vaccins anti-Escherichia coli , Escherichia coli pathogènes extra-intestinales , Hémolysines , Animaux , Escherichia coli pathogènes extra-intestinales/génétique , Escherichia coli pathogènes extra-intestinales/immunologie , Infections à Escherichia coli/prévention et contrôle , Infections à Escherichia coli/microbiologie , Infections à Escherichia coli/immunologie , Souris , Hémolysines/immunologie , Hémolysines/génétique , Protéines Escherichia coli/génétique , Protéines Escherichia coli/immunologie , Vaccins anti-Escherichia coli/immunologie , Antigènes bactériens/immunologie , Antigènes bactériens/génétique , Femelle , Facteurs de virulence/génétique , Facteurs de virulence/immunologie , Systèmes de sécrétion de type V/immunologie , Systèmes de sécrétion de type V/génétique , Modèles animaux de maladie humaine , Humains
5.
Virus Res ; 325: 199048, 2023 02.
Article de Anglais | MEDLINE | ID: mdl-36681192

RÉSUMÉ

As antimicrobial resistance (AMR) continues to increase, the therapeutic use of phages has re-emerged as an attractive alternative. However, knowledge of phage resistance development and bacterium-phage interaction complexity are still not fully interpreted. In this study, two lytic T4-like and T7-like phage infecting model Escherichia coli strain C600 are selected, and host genetic determinants involved in phage susceptibility and resistance are also identified using TraDIS strategy. Isolation and identification of the lytic T7-like show that though it belongs to the phage T7 family, genes encoding replication and transcription protein exhibit high differences. The TraDIS results identify a huge number of previously unidentified genes involved in phage infection, and a subset (six in susceptibility and nine in resistance) are shared under pressure of the two kinds of lytic phage. Susceptible gene wbbL has the highest value and implies the important role in phage susceptibility. Importantly, two susceptible genes QseE (QseE/QseF) and RstB (RstB/RstA), encoding the similar two-component system sensor histidine kinase (HKs), also identified. Conversely and strangely, outer membrane protein gene ompW, unlike the gene ompC encoding receptor protein of T4 phage, was shown to provide phage resistance. Overall, this study exploited a genome-wide fitness assay to uncover susceptibility and resistant genes, even the shared genes, important for the E. coli strain of both most popular high lytic T4-like and T7-like phages. This knowledge of the genetic determinants can be further used to analysis the behind function signatures to screen the potential agents to aid phage killing of MDR pathogens, which will greatly be valuable in improving the phage therapy outcome in fighting with microbial resistance.


Sujet(s)
Protéines Escherichia coli , Escherichia coli , Interactions hôte-microbes , Phages T , Bactériophage T7/génétique , Bactériophage T7/immunologie , Protéines de liaison à l'ADN , Escherichia coli/génétique , Escherichia coli/immunologie , Protéines Escherichia coli/génétique , Protéines Escherichia coli/immunologie , Récepteurs adrénergiques , Bactériophage T4/génétique , Bactériophage T4/immunologie , Phages T/génétique , Phages T/immunologie , Interactions hôte-microbes/génétique , Interactions hôte-microbes/immunologie
6.
Front Cell Infect Microbiol ; 12: 941939, 2022.
Article de Anglais | MEDLINE | ID: mdl-35967844

RÉSUMÉ

Lymphostatin is a virulence factor of enteropathogenic E. coli (EPEC) and non-O157 serogroup enterohaemorrhagic E. coli. Previous studies using whole-cell lysates of EPEC showed that lymphostatin inhibits the mitogen-activated proliferation of bulk human peripheral blood mononuclear cells (PBMCs) and the production of cytokines IL-2, IL-4, IL-5, and IFN-γ. Here, we used highly purified lymphostatin and PBMC-derived T cells to show that lymphostatin inhibits anti-CD3/anti-CD28-activated proliferation of human CD4+ and CD8+ T cells and blocks the synthesis of IL-2, IL-4, IL-10 and IFN-γ without affecting cell viability and in a manner dependent on an N-terminal DTD glycosyltransferase motif. Such inhibition was not observed with T cells activated by phorbol 12-myristate 13-acetate and ionomycin, implying that lymphostatin targets T cell receptor signaling. Analysis of the expression of CD69 indicated that lymphostatin suppresses T cell activation at an early stage and no impacts on apoptosis or necrosis were observed. Flow cytometric analysis of the DNA content of lymphostatin-treated CD4+ and CD8+ T cells showed a concentration- and DTD-dependent accumulation of the cells in the G0/G1 phase of the cell cycle, and corresponding reduction of the percentage of cells in S phase. Consistent with this, we found a marked reduction in the abundance of cyclins D3, E and A and loss of phosphorylated Rb over time in activated T cells from 8 donors treated with lymphostatin. Moreover, the cyclin-dependent kinase (cdk) inhibitor p27kip1, which inhibits progression of the cell cycle at G1 by acting on cyclin E-cdk2 or cyclin D-cdk4 complexes, was found to be accumulated in lymphostatin-treated T cells. Analysis of the abundance of phosphorylated kinases involved in signal transduction found that 30 of 39 were reduced in abundance following lymphostatin treatment of T cells from 5 donors, albeit not significantly so. Our data provide novel insights into the mode of action of lymphostatin on human T lymphocytes.


Sujet(s)
Toxines bactériennes , Infections à Escherichia coli , Protéines Escherichia coli , Escherichia coli , Lymphocytes T , Apoptose , Toxines bactériennes/immunologie , Lymphocytes T CD8+/immunologie , Points de contrôle du cycle cellulaire/immunologie , Division cellulaire , Prolifération cellulaire/physiologie , Cytokines/biosynthèse , Cytokines/immunologie , Escherichia coli entéropathogène/immunologie , Escherichia coli entéropathogène/pathogénicité , Escherichia coli/immunologie , Escherichia coli/pathogénicité , Infections à Escherichia coli/immunologie , Protéines Escherichia coli/immunologie , Humains , Interleukine-2 , Interleukine-4 , Agranulocytes/immunologie , Nécrose , Lymphocytes T/immunologie , Facteurs de virulence/immunologie
7.
Int J Mol Sci ; 23(2)2022 Jan 14.
Article de Anglais | MEDLINE | ID: mdl-35055067

RÉSUMÉ

α-hemolysin (HlyA) of E. coli binds irreversibly to human erythrocytes and induces cell swelling, ultimately leading to hemolysis. We characterized the mechanism involved in water transport induced by HlyA and analyzed how swelling and hemolysis might be coupled. Osmotic water permeability (Pf) was assessed by stopped-flow light scattering. Preincubation with HlyA strongly reduced Pf in control- and aquaporin 1-null red blood cells, although the relative Pf decrease was similar in both cell types. The dynamics of cell volume and hemolysis on RBCs was assessed by electrical impedance, light dispersion and hemoglobin release. Results show that HlyA induced erythrocyte swelling, which is enhanced by purinergic signaling, and is coupled to osmotic hemolysis. We propose a mathematical model of HlyA activity where the kinetics of cell volume and hemolysis in human erythrocytes depend on the flux of osmolytes across the membrane, and on the maximum volume that these cells can tolerate. Our results provide new insights for understanding signaling and cytotoxicity mediated by HlyA in erythrocytes.


Sujet(s)
Taille de la cellule , Érythrocytes/cytologie , Érythrocytes/physiologie , Protéines Escherichia coli/pharmacologie , Hémolysines/pharmacologie , Modèles biologiques , Adénosine triphosphate/métabolisme , Marqueurs biologiques , Mort cellulaire/effets des médicaments et des substances chimiques , Mort cellulaire/immunologie , Relation dose-effet des médicaments , Protéines Escherichia coli/immunologie , Hémolysines/immunologie , Hémolyse , Interactions hôte-pathogène , Humains , Cinétique , Perméabilité
8.
J Vet Sci ; 23(1): e7, 2022 Jan.
Article de Anglais | MEDLINE | ID: mdl-34841745

RÉSUMÉ

BACKGROUND: Enterotoxigenic Escherichia coli (ETEC) infection is a primary cause of livestock diarrhea. Therefore, effective vaccines are needed to reduce the incidence of ETEC infection. OBJECTIVES: Our study aimed to develop a multivalent ETEC vaccine targeting major virulence factors of ETEC, including enterotoxins and fimbriae. METHODS: SLS (STa-LTB-STb) recombinant enterotoxin and fimbriae proteins (F4, F5, F6, F18, and F41) were prepared to develop a multivalent vaccine. A total of 65 mice were immunized subcutaneously by vaccines and phosphate-buffered saline (PBS). The levels of specific immunoglobulin G (IgG) and pro-inflammatory cytokines were determined at 0, 7, 14 and 21 days post-vaccination (dpv). A challenge test with a lethal dose of ETEC was performed, and the survival rate of the mice in each group was recorded. Feces and intestine washes were collected to measure the concentrations of secretory immunoglobulin A (sIgA). RESULTS: Anti-SLS and anti-fimbriae-specific IgG in serums of antigen-vaccinated mice were significantly higher than those of the control group. Immunization with the SLS enterotoxin and multivalent vaccine increased interleukin-1ß (IL-1ß) and tumor necrosis factor-α (TNF-α) concentrations. Compared to diarrheal symptoms and 100% death of mice in the control group, mice inoculated with the multivalent vaccine showed an 80% survival rate without any symptom of diarrhea, while SLS and fimbriae vaccinated groups showed 60 and 70% survival rates, respectively. CONCLUSIONS: Both SLS and fimbriae proteins can serve as vaccine antigens, and the combination of these two antigens can elicit stronger immune responses. The results suggest that the multivalent vaccine can be successfully used for preventing ETEC in important livestock.


Sujet(s)
Diarrhée , Infections à Escherichia coli , Vaccins anti-Escherichia coli/immunologie , Animaux , Anticorps antibactériens/sang , Diarrhée/prévention et contrôle , Diarrhée/médecine vétérinaire , Modèles animaux de maladie humaine , Escherichia coli entérotoxigène , Entérotoxines/immunologie , Infections à Escherichia coli/prévention et contrôle , Infections à Escherichia coli/médecine vétérinaire , Protéines Escherichia coli/immunologie , Protéines de fimbriae/immunologie , Immunoglobuline A/analyse , Immunoglobuline G/sang , Souris , Vaccins combinés/immunologie
9.
Front Immunol ; 12: 753371, 2021.
Article de Anglais | MEDLINE | ID: mdl-34721427

RÉSUMÉ

Many pathogens enter the host via the gut, causing disease in animals and humans. A robust intestinal immune response is necessary to protect the host from these gut pathogens. Despite being best suited for eliciting intestinal immunity, oral vaccination remains a challenge due to the gastrointestinal environment, a poor uptake of vaccine antigens by the intestinal epithelium and the tolerogenic environment pervading the gut. To improve uptake, efforts have focused on targeting antigens towards the gut mucosa. An interesting target is aminopeptidase N (APN), a conserved membrane protein present on small intestinal epithelial cells shown to mediate epithelial transcytosis. Here, we aimed to further optimize this oral vaccination strategy in a large animal model. Porcine APN-specific monoclonal antibodies were generated and the most promising candidate in terms of epithelial transcytosis was selected to generate antibody fusion constructs, comprising a murine IgG1 or porcine IgA backbone and a low immunogenic antigen: the F18-fimbriated E. coli tip adhesin FedF. Upon oral delivery of these recombinant antibodies in piglets, both mucosal and systemic immune responses were elicited. The presence of the FedF antigen however appeared to reduce these immune responses. Further analysis showed that F18 fimbriae were able to disrupt the antigen presenting capacity of intestinal antigen presenting cells, implying potential tolerogenic effects of FedF. Altogether, these findings show that targeted delivery of molecules to epithelial aminopeptidase N results in their transcytosis and delivery to the gut immune systems. The results provide a solid foundation for the development of oral subunit vaccines to protect against gut pathogens.


Sujet(s)
Adhésines bactériennes/immunologie , Anticorps monoclonaux/immunologie , Antigènes bactériens/immunologie , Antigènes CD13/immunologie , Protéines Escherichia coli/immunologie , Immunoconjugués/immunologie , Immunoglobuline A/biosynthèse , Muqueuse intestinale/immunologie , Intestin grêle/immunologie , Suidae/immunologie , Transcytose , Vaccins synthétiques/immunologie , Adhésines bactériennes/administration et posologie , Administration par voie orale , Animaux , Anticorps antibactériens/biosynthèse , Anticorps antibactériens/immunologie , Anticorps monoclonaux/administration et posologie , Affinité des anticorps , Cellules présentatrices d'antigène/immunologie , Antigènes bactériens/administration et posologie , Antigènes CD13/physiologie , Escherichia coli entérotoxigène/immunologie , Cellules épithéliales/métabolisme , Protéines Escherichia coli/administration et posologie , Femelle , Fimbriae bactériens/immunologie , Immunoconjugués/administration et posologie , Immunoglobuline A/administration et posologie , Immunoglobuline A/immunologie , Immunoglobuline G/immunologie , Intestin grêle/enzymologie , Souris , Protéines de fusion recombinantes/administration et posologie , Protéines de fusion recombinantes/immunologie , Transcytose/physiologie , Vaccination/médecine vétérinaire
10.
Cell Host Microbe ; 29(10): 1482-1495.e12, 2021 10 13.
Article de Anglais | MEDLINE | ID: mdl-34582782

RÉSUMÉ

CRISPR-Cas systems provide immunity to bacteria by programing Cas nucleases with RNA guides that recognize and cleave infecting viral genomes. Bacteria and their viruses each encode recombination systems that could repair the cleaved viral DNA. However, it is unknown whether and how these systems can affect CRISPR immunity. Bacteriophage λ uses the Red system (gam-exo-bet) to promote recombination between related phages. Here, we show that λ Red also mediates evasion of CRISPR-Cas targeting. Gam inhibits the host E. coli RecBCD recombination system, allowing recombination and repair of the cleaved DNA by phage Exo-Beta, which promotes the generation of mutations within the CRISPR target sequence. Red recombination is strikingly more efficient than the host's RecBCD-RecA in the production of large numbers of phages that escape CRISPR targeting. These results reveal a role for Red-like systems in the protection of bacteriophages against sequence-specific nucleases, which may facilitate their spread across viral genomes.


Sujet(s)
Bactériophage lambda/génétique , Systèmes CRISPR-Cas , Escherichia coli/génétique , Mutation , Recombinaison génétique , Bactériophage lambda/immunologie , Bactériophage lambda/physiologie , Escherichia coli/immunologie , Escherichia coli/virologie , Protéines Escherichia coli/génétique , Protéines Escherichia coli/immunologie , Exodeoxyribonuclease V/génétique , Exodeoxyribonuclease V/immunologie , Interactions hôte-pathogène , Protéines virales/génétique , Protéines virales/immunologie
11.
mBio ; 12(3): e0017321, 2021 06 29.
Article de Anglais | MEDLINE | ID: mdl-34154415

RÉSUMÉ

Bacteria that colonize animals must overcome, or coexist, with the reactive oxygen species products of inflammation, a front-line defense of innate immunity. Among these is the neutrophilic oxidant bleach, hypochlorous acid (HOCl), a potent antimicrobial that plays a primary role in killing bacteria through nonspecific oxidation of proteins, lipids, and DNA. Here, we report that in response to increasing HOCl levels, Escherichia coli regulates biofilm production via activation of the diguanylate cyclase DgcZ. We identify the mechanism of DgcZ sensing of HOCl to be direct oxidation of its regulatory chemoreceptor zinc-binding (CZB) domain. Dissection of CZB signal transduction reveals that oxidation of the conserved zinc-binding cysteine controls CZB Zn2+ occupancy, which in turn regulates the catalysis of c-di-GMP by the associated GGDEF domain. We find DgcZ-dependent biofilm formation and HOCl sensing to be regulated in vivo by the conserved zinc-coordinating cysteine. Additionally, point mutants that mimic oxidized CZB states increase total biofilm. A survey of bacterial genomes reveals that many pathogenic bacteria that manipulate host inflammation as part of their colonization strategy possess CZB-regulated diguanylate cyclases and chemoreceptors. Our findings suggest that CZB domains are zinc-sensitive regulators that allow host-associated bacteria to perceive host inflammation through reactivity with HOCl. IMPORTANCE Immune cells are well equipped to eliminate invading bacteria, and one of their primary tools is the synthesis of bleach, hypochlorous acid (HOCl), the same chemical used as a household disinfectant. In this work, we present findings showing that many host-associated bacteria possess a bleach-sensing protein that allows them to adapt to the presence of this chemical in their environment. We find that the bacterium Escherichia coli responds to bleach by hunkering down and producing a sticky matrix known as biofilm, which helps it aggregate and adhere to surfaces. This behavior may play an important role in pathogenicity for E. coli and other bacteria, as it allows the bacteria to detect and adapt to the weapons of the host immune system.


Sujet(s)
Adhérence bactérienne/génétique , Biofilms/croissance et développement , GMP cyclique/analogues et dérivés , Protéines Escherichia coli/génétique , Escherichia coli/génétique , Régulation de l'expression des gènes bactériens , Inflammation/génétique , Transduction du signal , Bactéries/classification , Bactéries/génétique , Bactéries/immunologie , Bactéries/métabolisme , Adhérence bactérienne/immunologie , Biofilms/effets des médicaments et des substances chimiques , GMP cyclique/génétique , GMP cyclique/métabolisme , Escherichia coli/effets des médicaments et des substances chimiques , Escherichia coli/immunologie , Escherichia coli/métabolisme , Protéines Escherichia coli/immunologie , Protéines Escherichia coli/métabolisme , Génome bactérien , Acide hypochloreux/pharmacologie , Inflammation/immunologie
12.
Microbiology (Reading) ; 167(5)2021 05.
Article de Anglais | MEDLINE | ID: mdl-34032565

RÉSUMÉ

Bacteria often evolve resistance to phage through the loss or modification of cell surface receptors. In Escherichia coli and phage λ, such resistance can catalyze a coevolutionary arms race focused on host and phage structures that interact at the outer membrane. Here, we analyse another facet of this arms race involving interactions at the inner membrane, whereby E. coli evolves mutations in mannose permease-encoding genes manY and manZ that impair λ's ability to eject its DNA into the cytoplasm. We show that these man mutants arose concurrently with the arms race at the outer membrane. We tested the hypothesis that λ evolved an additional counter-defence that allowed them to infect bacteria with deleted man genes. The deletions severely impaired the ancestral λ, but some evolved phage grew well on the deletion mutants, indicating that they regained infectivity by evolving the ability to infect hosts independently of the mannose permease. This coevolutionary arms race fulfils the model of an inverse gene-for-gene infection network. Taken together, the interactions at both the outer and inner membranes reveal that coevolutionary arms races can be richer and more complex than is often appreciated.


Sujet(s)
Membrane bactérienne externe/immunologie , Bactériophage lambda/physiologie , Évolution biologique , Protéines Escherichia coli/immunologie , Escherichia coli/génétique , Escherichia coli/virologie , Membrane bactérienne externe/virologie , Bactériophage lambda/génétique , Escherichia coli/immunologie , Protéines Escherichia coli/génétique , Interactions hôte-pathogène , Mutation , Phosphoenolpyruvate-fructose phosphotransferase/génétique , Phosphoenolpyruvate-fructose phosphotransferase/immunologie
13.
Front Immunol ; 12: 654872, 2021.
Article de Anglais | MEDLINE | ID: mdl-34054818

RÉSUMÉ

Enhancement of mucosal immune responses in children and infants using novel adjuvants such as double mutant heat labile toxin (dmLT) is an important goal in the enteric vaccine field. dmLT has been shown to enhance mucosal IgA responses to the oral inactivated enterotoxigenic Escherichia coli (ETEC) vaccine ETVAX. dmLT can enhance IL-17A production from adult T cells, which may increase the production and secretion of mucosal IgA antibodies. However, the adjuvant mechanism remains to be fully elucidated and might differ between infants and adults due to age-related differences in the development of the immune system. The main objective of this study was to determine how dmLT influences antigen presenting cells and T cells from infants compared to adults, and the role of IL-1ß for mediating the adjuvant activity. Peripheral blood mononuclear cells (PBMCs) from Bangladeshi infants (6-11 months) and adults (18-40 years) were stimulated with the mitogen phytohaemagglutinin (PHA), the superantigen Staphylococcal enterotoxin B (SEB), ETVAX whole cell component (WCC) or E. coli lipopolysaccharide (LPS) ± dmLT, and cytokine production was measured using ELISA and electrochemiluminescence assays. The adjuvant dmLT significantly enhanced SEB- and PHA-induced IL-17A, but not IFN-γ responses, in PBMCs from both infants and adults. Blocking experiments using an IL-1 receptor antagonist demonstrated the importance of IL-1 signaling for the adjuvant effect. dmLT, ETVAX WCC and LPS induced dose-dependent IL-1ß responses of comparable magnitudes in infant and adult cells. Depletion experiments suggested that IL-1ß was mainly produced by monocytes. dmLT enhanced IL-1ß responses to low doses of WCC and LPS, and the adjuvant effect appeared over a wider dose-range of WCC in infants. dmLT and WCC also induced IL-6, IL-23 and IL-12p70 production in both age groups and dmLT tended to particularly enhance IL-23 responses to WCC. Our results show that dmLT can induce IL-1ß as well as other cytokines, which in turn may enhance IL-17A and potentially modulate other immunological responses in both infants and adults. Thus, dmLT may have an important function in promoting immune responses to the ETVAX vaccine, as well as other whole cell- or LPS-based vaccines in infants in low- and middle-income countries.


Sujet(s)
Adjuvants immunologiques , Antigènes bactériens/immunologie , Cytokines/biosynthèse , Lipopolysaccharides/immunologie , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Vaccins inactivés/immunologie , Adulte , Facteurs âges , Toxines bactériennes/immunologie , Entérotoxines/immunologie , Protéines Escherichia coli/immunologie , Humains , Nourrisson , Agranulocytes/immunologie , Agranulocytes/métabolisme , Activation des lymphocytes , Monocytes/immunologie , Monocytes/métabolisme , Jeune adulte
14.
Int Immunopharmacol ; 96: 107638, 2021 Jul.
Article de Anglais | MEDLINE | ID: mdl-33848909

RÉSUMÉ

The majority of urinary tract infections (UTIs) are caused by uropathogenic Escherichia coli (UPEC). Designing a vaccine will certainly reduce the occurrence of infection and antibiotic resistance of the isolates. Antigen 43 (Ag43) and autotransporter H (UpaH) have been associated with the virulence of UPEC. In the present study, the efficacy of different formulations of a hybrid protein composed of Ag43 and UpaH with and without alum and 1,25(OH)2D3 (Vitamin D3) adjuvants were evaluated in mice model. A significant increase in IgG and cellular responses was developed against Ag43::UpaH as compared to the control mice. The addition of alum or a mixture of alum and Vitamin D3 to the protein significantly enhanced the serum IgG responses and tended to remain in a steady state until 6 months. In addition, the mentioned formulations produced significant amounts of IgG1, IL-4, and IL-17 as compared to the fusion protein alone. In addition to the mentioned formulations, the combination of protein with Vitamin D3 also resulted in significantly higher serum IgA and IFN-γ levels as compared to the fusion protein alone. Mice immunized with fusion plus alum and formulation protein admixed with both alum and Vitamin D3 significantly reduced the bacterial load in the bladders and kidneys of mice as compared to the control. In this study, for the first time, the ability of a novel hybrid protein in combination with adjuvants alum and Vitamin D3 was evaluated against UPEC. Our results indicated that fusion Ag43::UpaH admixed with alum and Vitamin D3 could be a promising candidate against UTIs.


Sujet(s)
Alun/pharmacologie , Cholécalciférol/pharmacologie , Protéines Escherichia coli/immunologie , Protéines de fusion recombinantes/immunologie , Infections urinaires/prévention et contrôle , Escherichia coli uropathogène/immunologie , Facteurs de virulence/immunologie , Adjuvants immunologiques/pharmacologie , Adjuvants immunologiques/usage thérapeutique , Alun/administration et posologie , Animaux , Anticorps antibactériens/sang , Anticorps antibactériens/urine , Antigènes bactériens/immunologie , Charge bactérienne/effets des médicaments et des substances chimiques , Charge bactérienne/immunologie , Vaccins antibactériens/administration et posologie , Vaccins antibactériens/immunologie , Vaccins antibactériens/isolement et purification , Cholécalciférol/administration et posologie , Cytokines/métabolisme , Immunité humorale/effets des médicaments et des substances chimiques , Immunisation/méthodes , Immunoglobuline G/sang , Immunoglobuline G/urine , Injections veineuses , Souris de lignée BALB C , Muqueuse/immunologie , Protéines de fusion recombinantes/administration et posologie , Protéines de fusion recombinantes/isolement et purification , Infections urinaires/immunologie
15.
Infect Immun ; 89(7): e0010621, 2021 06 16.
Article de Anglais | MEDLINE | ID: mdl-33875477

RÉSUMÉ

There are no vaccines licensed for enterotoxigenic Escherichia coli (ETEC), a leading cause of diarrhea for children in developing countries and international travelers. Virulence heterogeneity among strains and difficulties identifying safe antigens for protective antibodies against STa, a potent but poorly immunogenic heat-stable toxin which plays a key role in ETEC diarrhea, are challenges in ETEC vaccine development. To overcome these challenges, we applied a toxoid fusion strategy and a novel epitope- and structure-based multiepitope fusion antigen (MEFA) vaccinology platform to construct two chimeric multivalent proteins, toxoid fusion 3xSTaN12S-mnLTR192G/L211A and adhesin CFA/I/II/IV MEFA, and demonstrated that the proteins induced protective antibodies against STa and heat-labile toxin (LT) produced by all ETEC strains or the seven most important ETEC adhesins (CFA/I and CS1 to CS6) expressed by the ETEC strains causing 60 to 70% of diarrheal cases and moderate to severe cases. Combining two proteins, we prepared a protein-based multivalent ETEC vaccine, MecVax. MecVax was broadly immunogenic; mice and pigs intramuscularly immunized with MecVax developed no apparent adverse effects but had robust antibody responses to the target toxins and adhesins. Importantly, MecVax-induced antibodies were broadly protective, demonstrated by significant adherence inhibition against E. coli bacteria producing any of the seven adhesins and neutralization of STa and cholera toxin (CT) enterotoxicity. Moreover, MecVax protected against watery diarrhea and provided over 70% and 90% protection against any diarrhea from an STa-positive or an LT-positive ETEC strain in a pig challenge model. These results indicated that MecVax induces broadly protective antibodies and prevents diarrhea preclinically, signifying that MecVax is potentially an effective injectable vaccine for ETEC. IMPORTANCE Enterotoxigenic Escherichia coli (ETEC) bacteria are a top cause of children's diarrhea and travelers' diarrhea and are responsible for over 220 million diarrheal cases and more than 100,000 deaths annually. A safe and effective ETEC vaccine can significantly improve public health, particularly in developing countries. Data from this preclinical study showed that MecVax induces broadly protective antiadhesin and antitoxin antibodies, becoming the first ETEC vaccine candidate to induce protective antibodies inhibiting adherence of the seven most important ETEC adhesins and neutralizing the enterotoxicity of not only LT but also STa toxin. More importantly, MecVax is shown to protect against clinical diarrhea from STa-positive or LT-positive ETEC infection in a pig challenge model, recording protection from antibodies induced by the protein-based, injectable, subunit vaccine MecVax against ETEC diarrhea and perhaps the possibility of intramuscularly administered protein vaccines for protection against intestinal mucosal infection.


Sujet(s)
Diarrhée/microbiologie , Diarrhée/prévention et contrôle , Escherichia coli entérotoxigène/immunologie , Infections à Escherichia coli/prévention et contrôle , Vaccins anti-Escherichia coli/immunologie , Animaux , Anticorps antibactériens/immunologie , Anticorps neutralisants/immunologie , Antigènes bactériens/immunologie , Toxines bactériennes/immunologie , Diarrhée/immunologie , Modèles animaux de maladie humaine , Épitopes/immunologie , Protéines Escherichia coli/immunologie , Vaccins anti-Escherichia coli/administration et posologie , Vaccins anti-Escherichia coli/effets indésirables , Souris , Protéines de fusion recombinantes/immunologie , Suidae , Vaccins combinés/génétique , Vaccins combinés/immunologie
16.
J Pharm Pharmacol ; 73(2): 135-144, 2021 Mar 04.
Article de Anglais | MEDLINE | ID: mdl-33793799

RÉSUMÉ

OBJECTIVES: Propolis is a bee-made product used for centuries due to its diverse biological properties, including its immunomodulatory action. This work aimed at investigating whether propolis may affect monocyte functions challenged with retinoic acid (RA), B subunit of Escherichia coli heat-labile enterotoxin (EtxB), human melanoma-associated antigen-1 (MAGE-1) and lipopolysaccharide (LPS). METHODS: Monocytes from healthy donors were treated with the stimuli separately or in the presence of propolis. Cell viability was evaluated by MTT assay, cell marker expression was assessed by flow cytometry, cytokine production by ELISA, gene expression by RT-qPCR. KEY FINDINGS: Propolis alone maintained TLR-2, TLR-4, HLA-DR, CD40 and CD80 expression in the monocytes; however, its combination with either MAGE-1 or LPS decreased CD40 expression triggered by the stimuli. Propolis maintained RA action on cell marker expression. Propolis inhibited TNF-α (with either EtxB or MAGE-1) and IL-6 (with either RA or MAGE-1), and increased IL-10 (with MAGE-1) production. Propolis downmodulated LC3 expression induced by LPS. It also induced a lower NF-kB expression than control cells and its combination with RA induced a higher expression than the stimulus alone. CONCLUSIONS: Propolis potentially affected innate immunity by downmodulating the monocytes pro-inflammatory activity.


Sujet(s)
Cytokines/métabolisme , Immunité innée/effets des médicaments et des substances chimiques , Monocytes/effets des médicaments et des substances chimiques , Propolis/pharmacologie , Adulte , Animaux , Toxines bactériennes/immunologie , Abeilles , Marqueurs biologiques/métabolisme , Brésil , Survie cellulaire/effets des médicaments et des substances chimiques , Entérotoxines/immunologie , Protéines Escherichia coli/immunologie , Humains , Monocytes/immunologie , Facteur de transcription NF-kappa B/métabolisme , Trétinoïne/pharmacologie
17.
Mol Immunol ; 134: 218-227, 2021 06.
Article de Anglais | MEDLINE | ID: mdl-33823320

RÉSUMÉ

Enterohemorrhagic Escherichia coli (EHEC) O157:H7 strain is known as one of the major human foodborne pathogens. Lack of effective clinical treatment for human diarrheal diseases confirms the need for vaccine production against enteric bacteria such as E.coli O157:H7. Shiga-like toxin (Stx), EscC, and Intimin are the main important virulent factors of this enteric pathogen. In the present study, a comparative Omics analysis was conducted to identify most invasion EHEC antigenic factors as a potential immunogen. SEI (Stx-EscC-Intimin) trivalent chimeric protein was designed from the exposed and epitope rich part of these virulence factors. Sequence optimization, physicochemical properties, mRNA folding, three-dimensional structure and immunoinformatics data were investigated. The chimeric gene was synthesized with codon bias of E. coli. Recombinant protein was expressed and confirmed by western blot analysis. To evaluate the immunogenicity of the designed protein, the protein was administered to BALB/c mice and the serum IgG was determined by ELISA. Based on the Ramachandran plot, the validation data showed that 90.1 % of residues lie in the favored region. The high antigenicity of the multimeric protein was predicted by the immunoinformatic analysis. Epitope prediction had shown the proper distribution of linear and conformational B-cell epitopes and the competition of T-cell epitopes to bind MHC molecules too. Recombinant ESI Protein with 74.5 kDa was expressed in E. coli. Western blot analysis by anti-Stx antibody, confirmed a single band of chimeric protein. Consequently, the chimeric gene was designed and constructed after assessments. From in silico approach, the protein deduced from this cassette can be an immunogen candidate, and act against toxicity and adherence of EHEC.


Sujet(s)
Adhésines bactériennes/immunologie , Infections à Escherichia coli , Protéines Escherichia coli/immunologie , Vaccins anti-Escherichia coli/immunologie , Protéines de fusion recombinantes/immunologie , Shiga-toxines/immunologie , Systèmes de sécrétion de type III/immunologie , Animaux , Anticorps antibactériens/immunologie , Adhérence bactérienne , Biologie informatique , Infections à Escherichia coli/immunologie , Infections à Escherichia coli/prévention et contrôle , Escherichia coli O157/immunologie , Femelle , Gènes bactériens/immunologie , Souris , Souris de lignée BALB C
18.
Gut Microbes ; 13(1): 1-13, 2021.
Article de Anglais | MEDLINE | ID: mdl-33645430

RÉSUMÉ

Recent studies have gained a better appreciation of the potential impacts of enteric infections beyond symptomatic diarrhea. It is recognized that infections by several enteropathogens could be associated with growth deficits in children and intestinal and systemic inflammation may play an important underlying role. With enterotoxigenic E. coli (ETEC) being one of the leading causes of diarrhea among children in the developing world and important contributor to stunting, a better understanding of the impact of ETEC infection beyond diarrhea is timely and greatly needed. To address this, we evaluated if ETEC infection induces intestinal and systemic inflammation and its impact on colonization and immune responses to ETEC vaccine-specific antigens in a dose descending experimental human challenge model using ETEC strain H10407. This study demonstrates that the concentrations of myeloperoxidase (MPO) in stool and intestinal fatty acid-binding protein (an indicator of compromised intestinal epithelial integrity) in serum, significantly increased following ETEC infection in both diarrhea and asymptomatic cases and the magnitudes and kinetics of MPO are dose and clinical outcome dependent. Cytokines IL-17A and IFN-γ were significantly increased in serum post-ETEC challenge. In addition, higher pre-challenge concentrations of cytokines IL-10 and GM-CSF were associated with protection from ETEC diarrhea. Interestingly, higher MPO concentrations were associated with higher intestinal colonization of ETEC and lower seroconversions of colonization factor I antigen, but the reverse was noted for seroconversions to heat-labile toxin B-subunit. Together this study has important implications for understanding the acute and long-term negative health outcomes associated with ETEC infection.


Sujet(s)
Escherichia coli entérotoxigène/croissance et développement , Escherichia coli entérotoxigène/immunologie , Infections à Escherichia coli/immunologie , Infections à Escherichia coli/microbiologie , Inflammation/microbiologie , Intestins/microbiologie , Anticorps antibactériens/sang , Infections asymptomatiques , Toxines bactériennes/immunologie , Cytokines/sang , Diarrhée/microbiologie , Entérotoxines/immunologie , Protéines Escherichia coli/immunologie , Protéines de liaison aux acides gras/sang , Fèces/composition chimique , Protéines de fimbriae/immunologie , Humains , Immunoglobuline A/sang , Immunoglobuline G/sang , Inflammation/immunologie , Myeloperoxidase/analyse
19.
Methods Mol Biol ; 2291: 253-272, 2021.
Article de Anglais | MEDLINE | ID: mdl-33704757

RÉSUMÉ

Therapeutic antibodies (Abs) inhibiting bacterial adhesion to host epithelia are an attractive option to reduce the load of Shiga toxin-producing E. coli (STEC) in the intestine of the patient and also in the bovine reservoir, thereby minimizing the risk of STEC contamination in the food chain. Of particular interest are recombinant single-domain Ab fragments called nanobodies (Nbs) derived from the variable domain of camelid heavy chain-only antibodies (VHH). The outer membrane adhesin intimin and the translocated intimin receptor (Tir) are essential for the attachment of STEC to host epithelia. In addition, EspA filaments of the bacterial type III protein secretion system are needed for Tir translocation into the host cell. Given their importance for bacterial adhesion and colonization, we developed Nbs against intimin, Tir and EspA proteins of STEC serotype O157:H7. Here, we report the screening methods used to isolate inhibitory Nbs blocking intimin-Tir protein-protein interaction, actin-pedestal formation, and intimate adhesion of STEC to epithelial cells in vitro. First, we describe how VHH gene repertoires can be produced as Nbs secreted by E. coli using the α-hemolysin (HlyA) protein secretion system. Next, we report the methods for identification of inhibitors of intimin-Tir protein-protein interaction and of STEC intimate adhesion to HeLa cells in culture. These methods can be adapted for the screening of Nbs against different adhesin-receptor complexes to block the adhesion of other pathogens to host cells.


Sujet(s)
Adhésines bactériennes/immunologie , Anticorps antibactériens/immunologie , Adhérence bactérienne/immunologie , Cellules épithéliales , Escherichia coli O157/immunologie , Protéines Escherichia coli/immunologie , Récepteurs de surface cellulaire/immunologie , Anticorps à domaine unique/immunologie , Animaux , Bovins , Cellules épithéliales/immunologie , Cellules épithéliales/microbiologie , Escherichia coli O157/pathogénicité , Humains
20.
Sci Rep ; 11(1): 3789, 2021 02 15.
Article de Anglais | MEDLINE | ID: mdl-33589691

RÉSUMÉ

The interactions of the antibiotic proteins colicins/pyocins with immunity proteins is a seminal model system for studying protein-protein interactions and specificity. Yet, a precise and quantitative determination of which structural elements and residues determine their binding affinity and specificity is still lacking. Here, we used comparative structure-based energy calculations to map residues that substantially contribute to interactions across native and engineered complexes of colicins/pyocins and immunity proteins. We show that the immunity protein α1-α2 motif is a unique structurally-dissimilar element that restricts interaction specificity towards all colicins/pyocins, in both engineered and native complexes. This motif combines with a diverse and extensive array of electrostatic/polar interactions that enable the exquisite specificity that characterizes these interactions while achieving ultra-high affinity. Surprisingly, the divergence of these contributing colicin residues is reciprocal to residue conservation in immunity proteins. The structurally-dissimilar immunity protein α1-α2 motif is recognized by divergent colicins similarly, while the conserved immunity protein α3 helix interacts with diverse colicin residues. Electrostatics thus plays a key role in setting interaction specificity across all colicins and immunity proteins. Our analysis and resulting residue-level maps illuminate the molecular basis for these protein-protein interactions, with implications for drug development and rational engineering of these interfaces.


Sujet(s)
Colicines/ultrastructure , Protéines de liaison à l'ADN/ultrastructure , Protéines Escherichia coli/ultrastructure , Pyocines/composition chimique , Protéines de liaison à l'ARN/ultrastructure , Séquence d'acides aminés/génétique , Sites de fixation/génétique , Colicines/composition chimique , Colicines/génétique , Colicines/immunologie , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/immunologie , Protéines Escherichia coli/composition chimique , Protéines Escherichia coli/génétique , Protéines Escherichia coli/immunologie , Complexes multiprotéiques/composition chimique , Complexes multiprotéiques/génétique , Complexes multiprotéiques/ultrastructure , Liaison aux protéines/génétique , Cartes d'interactions protéiques/génétique , Cartes d'interactions protéiques/immunologie , Structure secondaire des protéines , Pyocines/immunologie , Protéines de liaison à l'ARN/génétique , Protéines de liaison à l'ARN/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE